1
|
Orekhov AN, Ivanova EA, Markin AM, Nikiforov NG, Sobenin IA. Genetics of Arterial-Wall-Specific Mechanisms in Atherosclerosis: Focus on Mitochondrial Mutations. Curr Atheroscler Rep 2020; 22:54. [PMID: 32772280 DOI: 10.1007/s11883-020-00873-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Mutations in both nuclear and mitochondrial genes are associated with the development of atherosclerotic lesions in arteries and may provide a partial explanation to the focal nature of lesion distribution in the arterial wall. This review is aimed to discuss the genetic aspects of atherogenesis with a special focus on possible pro-atherogenic variants (mutations) of the nuclear and mitochondrial genomes that may be implicated in atherosclerosis development and progression. RECENT FINDINGS Mutations in the nuclear genes generally do not cause a phenotype restricted to a specific vascular wall cell and manifest themselves mostly at the organism level. Such mutations can act as important contributors to changes in lipid metabolism and modulate other risk factors of atherosclerosis. By contrast, mitochondrial DNA (mtDNA) mutations occurring locally in the arterial wall cells or in circulating immune cells may play a site-specific role in atherogenesis. The mosaic distribution of heteroplasmic mtDNA mutations in the arterial wall tissue may explain, at least to some extent, the locality and focality of atherosclerotic lesions distribution. The genetic mechanisms of atherogenesis include alterations of both nuclear and mitochondrial genomes. Altered lipid metabolism and inflammatory response of resident arterial wall and circulating immune cells may be related to mtDNA damage and defective mitophagy, which hinders clearance of dysfunctional mitochondria. Mutations of mtDNA can have mosaic distribution and locally affect functionality of endothelial and subendothelial intimal cells in the arterial wall contributing to atherosclerotic lesion development.
Collapse
Affiliation(s)
- Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315. .,Laboratory of Infection Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, Moscow, Russia, 117418.
| | - Ekaterina A Ivanova
- Institute for Atherosclerosis Research, 2-1-207 Osennyaya Street, Moscow, Russia, 121609.
| | - Alexander M Markin
- Laboratory of Infection Pathology and Molecular Microecology, Institute of Human Morphology, 3 Tsyurupa Street, Moscow, Russia, 117418
| | - Nikita G Nikiforov
- Centre of Collective Usage, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilova Street, Moscow, Russia, 119334.,Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, Moscow, Russia, 121552
| | - Igor A Sobenin
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow, Russia, 125315.,Laboratory of Medical Genetics, Institute of Experimental Cardiology, National Medical Research Center of Cardiology, 15A 3-rd Cherepkovskaya Street, Moscow, Russia, 121552
| |
Collapse
|
2
|
Singh S, Nguyen H, Michels D, Bazinet H, Matkar PN, Liu Z, Esene L, Adam M, Bugyei‐Twum A, Mebrahtu E, Joseph J, Ehsan M, Chen HH, Qadura M, Singh KK. BReast CAncer susceptibility gene 2 deficiency exacerbates oxidized LDL-induced DNA damage and endothelial apoptosis. Physiol Rep 2020; 8:e14481. [PMID: 32638521 PMCID: PMC7340845 DOI: 10.14814/phy2.14481] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/18/2020] [Accepted: 05/15/2020] [Indexed: 12/24/2022] Open
Abstract
Mutations in the tumor suppressor gene BRCA2 (BReast CAncer susceptibility gene 2) predispose carriers to breast, ovarian, and other cancers. In response to DNA damage, BRCA2 participates in homology-directed DNA damage repair to maintain genome stability. Genome-wide association studies have identified an association between BRCA2 single nucleotide polymorphisms and plasma-lipid levels and lipid deregulation in humans. To date, DNA damage, apoptosis, and lipid deregulation are recognized as central pathways for endothelial dysfunction and atherosclerosis; however, the role of BRCA2 in endothelial dysfunction remains to be elucidated. To determine the role of BRCA2 in endothelial dysfunction, BRCA2 was silenced in human umbilical vein endothelial cells (ECs) and assessed for markers of DNA damage, apoptosis, and endothelial function following oxidized low-density lipoprotein (oxLDL) treatment. OxLDL was found to induce significant reactive oxygen species (ROS) production in BRCA2-silenced ECs. This increase in ROS production was associated with exacerbated DNA damage evidenced by increased expression and activation of DNA double-stranded break (DSB) marker γH2AX and reduced RAD51-foci formation-an essential regulator of DSB repair. Increased DSBs were associated with enhanced expression and activation of pro-apoptotic p53 and significant apoptosis in oxLDL-treated BRCA2-silenced ECs. Loss of BRCA2 in ECs was further associated with oxLDL-induced impaired tube-forming potential and eNOS expression. Collectively, the data reveals, for the first time, a novel role of BRCA2 as a regulator of EC survival and function in the setting of oxLDL treatment in vitro. Additionally, the data provide important clues regarding the potential susceptibility of BRCA2 mutation carriers to endothelial dysfunction, atherosclerosis, and other cardiovascular diseases.
Collapse
Affiliation(s)
- Shweta Singh
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hien Nguyen
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - David Michels
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hannah Bazinet
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Pratiek N. Matkar
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Zongyi Liu
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Lilian Esene
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Mohamed Adam
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Antoinette Bugyei‐Twum
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Elizabeth Mebrahtu
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Jameela Joseph
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Department of BiologyUniversity of Western OntarioLondonONCanada
| | - Mehroz Ehsan
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hao H. Chen
- Division of CardiologyKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael's HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Mohammad Qadura
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Vascular SurgeryKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
| | - Krishna K. Singh
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Vascular SurgeryKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoONCanada
- Department of SurgeryUniversity of TorontoTorontoONCanada
- Pharmacology and ToxicologyUniversity of TorontoTorontoONCanada
| |
Collapse
|
3
|
Kress JM, Dio LD, Heck L, Pulliero A, Izzotti A, Laarmann K, Fritz G, Kaina B. Human primary endothelial cells are impaired in nucleotide excision repair and sensitive to benzo[a]pyrene compared with smooth muscle cells and pericytes. Sci Rep 2019; 9:13800. [PMID: 31551436 PMCID: PMC6760230 DOI: 10.1038/s41598-019-49953-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 08/30/2019] [Indexed: 11/24/2022] Open
Abstract
The endothelium represents the inner cell layer of blood vessels and is supported by smooth muscle cells and pericytes, which form the vessel structure. The endothelium is involved in the pathogenesis of many diseases, including the development of atherosclerosis. Due to direct blood contact, the blood vessel endothelium is inevitably exposed to genotoxic substances that are systemically taken up by the body, including benzo[a]pyrene, which is a major genotoxic component in cigarette smoke and a common environmental mutagen and human carcinogen. Here, we evaluated the impact of benzo[a]pyrene diol epoxide (BPDE), which is the reactive metabolite of benzo[a]pyrene, on the three innermost vessel cell types. Primary human endothelial cells (HUVEC), primary human smooth muscle cells (HUASMC) and primary human pericytes (HPC) were treated with BPDE, and analyses of cytotoxicity, cellular senescence and genotoxic effects were then performed. The results showed that HUVEC were more sensitive to the cytotoxic activity of BPDE than HUASMC and HPC. We further show that HUVEC display a detraction in the repair of BPDE-induced adducts, as determined through the comet assay and the quantification of BPDE adducts in post-labelling experiments. A screening for DNA repair factors revealed that the nucleotide excision repair (NER) proteins ERCC1, XPF and ligase I were expressed at lower levels in HUVEC compared with HUASMC and HPC, which corresponds with the impaired NER-mediated removal of BPDE adducts from DNA. Taken together, the data revealed that HUVEC exhibit an unexpected DNA repair-impaired phenotype, which has implications on the response of the endothelium to genotoxicants that induce bulky DNA lesions, including the development of vascular diseases resulting from smoking and environmental pollution.
Collapse
Affiliation(s)
- Joana M Kress
- Institute of Toxicology, University Medical Center, Mainz, 55131, Germany
| | - Lorella Di Dio
- Institute of Toxicology, University Medical Center, Mainz, 55131, Germany
| | - Larissa Heck
- Institute of Toxicology, University Medical Center, Mainz, 55131, Germany
| | - Alessandra Pulliero
- Department of Health Sciences, University of Genoa, Genoa, 16132, Italy.,Hospital Policlinico San Martino, University of Genoa, Genoa, 16132, Italy
| | - Alberto Izzotti
- Hospital Policlinico San Martino, University of Genoa, Genoa, 16132, Italy
| | - Kathrin Laarmann
- Institute of Toxicology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | - Bernd Kaina
- Institute of Toxicology, University Medical Center, Mainz, 55131, Germany.
| |
Collapse
|
4
|
Crocco P, Montesanto A, Dato S, Geracitano S, Iannone F, Passarino G, Rose G. Inter-Individual Variability in Xenobiotic-Metabolizing Enzymes: Implications for Human Aging and Longevity. Genes (Basel) 2019; 10:genes10050403. [PMID: 31137904 PMCID: PMC6562959 DOI: 10.3390/genes10050403] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/15/2019] [Accepted: 05/23/2019] [Indexed: 01/01/2023] Open
Abstract
Xenobiotic-metabolizing enzymes (XME) mediate the body’s response to potentially harmful compounds of exogenous/endogenous origin to which individuals are exposed during their lifetime. Aging adversely affects such responses, making the elderly more susceptible to toxics. Of note, XME genetic variability was found to impact the ability to cope with xenobiotics and, consequently, disease predisposition. We hypothesized that the variability of these genes influencing the interaction with the exposome could affect the individual chance of becoming long-lived. We tested this hypothesis by screening a cohort of 1112 individuals aged 20–108 years for 35 variants in 23 XME genes. Four variants in different genes (CYP2B6/rs3745274-G/T, CYP3A5/rs776746-G/A, COMT/rs4680-G/A and ABCC2/rs2273697-G/A) differently impacted the longevity phenotype. In particular, the highest impact was observed in the age group 65–89 years, known to have the highest incidence of age-related diseases. In fact, genetic variability of these genes we found to account for 7.7% of the chance to survive beyond the age of 89 years. Results presented herein confirm that XME genes, by mediating the dynamic and the complex gene–environment interactions, can affect the possibility to reach advanced ages, pointing to them as novel genes for future studies on genetic determinants for age-related traits.
Collapse
Affiliation(s)
- Paolina Crocco
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy.
| | - Alberto Montesanto
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy.
| | - Serena Dato
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy.
| | - Silvana Geracitano
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy.
| | - Francesca Iannone
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy.
| | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy.
| | - Giuseppina Rose
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy.
| |
Collapse
|
5
|
Schwartz SM, Virmani R, Majesky MW. An update on clonality: what smooth muscle cell type makes up the atherosclerotic plaque? F1000Res 2018; 7:F1000 Faculty Rev-1969. [PMID: 30613386 PMCID: PMC6305222 DOI: 10.12688/f1000research.15994.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
Almost 50 years ago, Earl Benditt and his son John described the clonality of the atherosclerotic plaque. This led Benditt to propose that the atherosclerotic lesion was a smooth muscle neoplasm, similar to the leiomyomata seen in the uterus of most women. Although the observation of clonality has been confirmed many times, interest in the idea that atherosclerosis might be a form of neoplasia waned because of the clinical success of treatments for hyperlipemia and because animal models have made great progress in understanding how lipid accumulates in the plaque and may lead to plaque rupture. Four advances have made it important to reconsider Benditt's observations. First, we now know that clonality is a property of normal tissue development. Second, this is even true in the vessel wall, where we now know that formation of clonal patches in that wall is part of the development of smooth muscle cells that make up the tunica media of arteries. Third, we know that the intima, the "soil" for development of the human atherosclerotic lesion, develops before the fatty lesions appear. Fourth, while the cells comprising this intima have been called "smooth muscle cells", we do not have a clear definition of cell type nor do we know if the initial accumulation is clonal. As a result, Benditt's hypothesis needs to be revisited in terms of changes in how we define smooth muscle cells and the quite distinct developmental origins of the cells that comprise the muscular coats of all arterial walls. Finally, since clonality of the lesions is real, the obvious questions are do these human tumors precede the development of atherosclerosis, how do the clones develop, what cell type gives rise to the clones, and in what ways do the clones provide the soil for development and natural history of atherosclerosis?
Collapse
Affiliation(s)
| | - Renu Virmani
- CV Path Institute, Gaithersberg, Maryland, 20878, USA
| | - Mark W. Majesky
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Hospital Research Institute, Seattle, WA, 98112, USA
| |
Collapse
|
6
|
Abstract
Ischaemic heart disease and stroke are vascular events with serious health consequences worldwide. Recent genetic and epigenetic techniques have revealed many genetic determinants of these vascular events and simplified the approaches to research focused on ischaemic heart disease and stroke. The pathogenetic mechanisms of ischaemic heart disease and stroke are complex, with mitochondrial involvement (partially or entirely) recently gaining substantial support. Not only can mitochondrial reactive oxygen species give rise to ischaemic heart disease and stroke by production of oxidised low-density lipoprotein and induction of apoptosis, but the impact on pericytes contributes directly to the pathogenesis. Over the past two decades, publications implicate the causative role of nuclear genes in the development of ischaemic heart disease and stroke, in contrast to the potential role of mitochondrial DNA (mtDNA) in the pathophysiology of the disorders, which is much less understood, although recent studies do demonstrate that the involvement of mitochondria and mtDNA in the development of ischaemic heart disease and stroke is likely to be larger than originally thought, with the novel discovery of links among mitochondria, mtDNA and vascular events. Here we explore the molecular events and mtDNA alterations in relation to the role of mitochondria in ischaemic heart disease and stroke.
Collapse
|
7
|
Environmental carcinogens and mutational pathways in atherosclerosis. Int J Hyg Environ Health 2015; 218:293-312. [DOI: 10.1016/j.ijheh.2015.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 01/26/2015] [Accepted: 01/29/2015] [Indexed: 02/07/2023]
|
8
|
Saccà SC, Roszkowska AM, Izzotti A. Environmental light and endogenous antioxidants as the main determinants of non-cancer ocular diseases. Mutat Res 2013; 752:153-171. [PMID: 23337404 DOI: 10.1016/j.mrrev.2013.01.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 01/10/2013] [Accepted: 01/11/2013] [Indexed: 02/03/2023]
Abstract
The human eye is constantly exposed to sunlight and artificial lighting. Exogenous sources of reactive oxygen species (ROS) such as UV light, visible light, ionizing radiation, chemotherapeutics, and environmental toxins contribute to oxidative damage in ocular tissues. Long-term exposure to these insults places the aging eye at considerable risk for pathological consequences of oxidative stress. Furthermore, in eye tissues, mitochondria are an important endogenous source of ROS. Over time, all ocular structures, from the tear film to the retina, undergo oxidative stress, and therefore, the antioxidant defenses of each tissue assume the role of a safeguard against degenerative ocular pathologies. The ocular surface and cornea protect the other ocular tissues and are significantly exposed to oxidative stress of environmental origin. Overwhelming of antioxidant defenses in these tissues clinically manifests as pathologies including pterygium, corneal dystrophies, and endothelial Fuch's dystrophy. The crystalline lens is highly susceptible to oxidative damage in aging because its cells and their intracellular proteins are not turned over or replaced, thus providing the basis for cataractogenesis. The trabecular meshwork, which is the anterior chamber tissue devoted to aqueous humor drainage, has a particular susceptibility to mitochondrial oxidative injury that affects its endothelium and leads to an intraocular pressure increase that marks the beginning of glaucoma. Photo-oxidative stress can cause acute or chronic retinal damage. The pathogenesis of age-related macular degeneration involves oxidative stress and death of the retinal pigment epithelium followed by death of the overlying photoreceptors. Accordingly, converging evidence indicates that mutagenic mechanisms of environmental and endogenous sources play a fundamental pathogenic role in degenerative eye diseases.
Collapse
Affiliation(s)
- Sergio C Saccà
- Department of Head/Neck Pathologies, St Martino Hospital, Ophthalmology unit, Genoa, Italy
| | - Anna Maria Roszkowska
- Department of Specialized Surgery, University Hospital, Ophthalmology Unit, Messina, Italy
| | - Alberto Izzotti
- Department of Health Sciences, University of Genoa, Via A. Pastore 1, I-16132, Genoa, Italy.
| |
Collapse
|
9
|
Cervelli T, Borghini A, Galli A, Andreassi MG. DNA damage and repair in atherosclerosis: current insights and future perspectives. Int J Mol Sci 2012; 13:16929-44. [PMID: 23443128 PMCID: PMC3546731 DOI: 10.3390/ijms131216929] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/20/2012] [Accepted: 12/05/2012] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is the leading cause of morbidity and mortality among Western populations. Over the past two decades, considerable evidence has supported a crucial role for DNA damage in the development and progression of atherosclerosis. These findings support the concept that the prolonged exposure to risk factors (e.g., dyslipidemia, smoking and diabetes mellitus) leading to reactive oxygen species are major stimuli for DNA damage within the plaque. Genomic instability at the cellular level can directly affect vascular function, leading to cell cycle arrest, apoptosis and premature vascular senescence. The purpose of this paper is to review current knowledge on the role of DNA damage and DNA repair systems in atherosclerosis, as well as to discuss the cellular response to DNA damage in order to shed light on possible strategies for prevention and treatment.
Collapse
Affiliation(s)
- Tiziana Cervelli
- Institute of Clinical Physiology, CNR (The National Research Council), via Moruzzi 1, 56124 Pisa, Italy.
| | | | | | | |
Collapse
|
10
|
Changes in hepatic protein expression in spontaneously hypertensive rats suggest early stages of non-alcoholic fatty liver disease. J Proteomics 2012; 75:1752-63. [DOI: 10.1016/j.jprot.2011.12.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Revised: 11/03/2011] [Accepted: 12/10/2011] [Indexed: 02/07/2023]
|
11
|
Abstract
Evidence for the association of DNA damage with cardiovascular disease has been obtained from in vitro cell culture models, experimental cardiovascular disease and analysis of samples obtained from humans with disease. There is general acceptance that several factors associated with the risk of developing cardiovascular disease cause oxidative damage to DNA in cell culture models with both nuclear and mitochondrial DNA as targets. Moreover, evidence obtained over the past 10 years points to a possible mechanistic role for DNA damage in experimental atherosclerosis culminating in recent studies challenging the assumption that DNA damage is merely a biomarker of the disease process. This kind of mechanistic insight provides a renewed impetus for further studies in this area.
Collapse
Affiliation(s)
- Qudsia Malik
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital , Leicester , UK
| | | |
Collapse
|
12
|
Taspinar M, Aydos S, Sakiragaoglu O, Duzen IV, Yalcinkaya A, Oztuna D, Bardakci H, Tutar E, Sunguroglu A. Impact of Genetic Variations of theCYP1A1, GSTT1, andGSTM1Genes on the Risk of Coronary Artery Disease. DNA Cell Biol 2012; 31:211-8. [DOI: 10.1089/dna.2011.1252] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Mehmet Taspinar
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Sena Aydos
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Onur Sakiragaoglu
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Irfan Veysel Duzen
- Department of Cardiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Adnan Yalcinkaya
- Department of Cardiovascular Surgery, Turkiye Yuksek Ihtisas Hospital, Ankara, Turkey
| | - Derya Oztuna
- Department of Biostatistics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Hasmet Bardakci
- Department of Cardiovascular Surgery, Turkiye Yuksek Ihtisas Hospital, Ankara, Turkey
| | - Eralp Tutar
- Department of Cardiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Asuman Sunguroglu
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
13
|
Abstract
Clinical and experimental studies in humans provide evidence that moderate physical activity significantly decreases artery oxidative damage to nuclear DNA, DNA-adducts related to age and dyslipedemia, and mitochondrial DNA damage. Maintenance of adequate mitochondrial function is crucial for preventing lipid accumulation and peroxidation occurring in atherosclerosis. Studies performed on human muscle biopsies analyzing gene expression in living humans reveal that physically active subjects improve the expression of genes involved in mitochondrial function and of related microRNAs. The attenuation of oxidative damage to nuclear and mitochondrial DNA by physical activity resulted in beneficial effects due to polymorphisms of glutathione S-transferases genes. Subjects bearing null GSTM1/T1 polymorphisms have poor life expectancy in the case of being sedentary, which was increased 2.6-fold in case they performed physical activity. These findings indicate that the preventive effect of physical activity undergoes interindividual variation affected by genetic polymorphisms.
Collapse
Affiliation(s)
- Alberto Izzotti
- Department of Health Sciences, Faculty of Medicine, University of Genoa, Genoa, Italy.
| |
Collapse
|
14
|
Ketelslegers HB, Godschalk RWL, Gottschalk RWH, Knaapen AM, Koppen G, Schoeters G, Baeyens WF, Nelen V, Geraedts JPM, van Delft JHM, Kleinjans JCS, van Larebeke NA. Prevalence of at-risk genotypes for genotoxic effects decreases with age in a randomly selected population in Flanders: a cross sectional study. Environ Health 2011; 10:85. [PMID: 21975123 PMCID: PMC3195704 DOI: 10.1186/1476-069x-10-85] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 10/05/2011] [Indexed: 05/03/2023]
Abstract
BACKGROUND We hypothesized that in Flanders (Belgium), the prevalence of at-risk genotypes for genotoxic effects decreases with age due to morbidity and mortality resulting from chronic diseases. Rather than polymorphisms in single genes, the interaction of multiple genetic polymorphisms in low penetrance genes involved in genotoxic effects might be of relevance. METHODS Genotyping was performed on 399 randomly selected adults (aged 50-65) and on 442 randomly selected adolescents. Based on their involvement in processes relevant to genotoxicity, 28 low penetrance polymorphisms affecting the phenotype in 19 genes were selected (xenobiotic metabolism, oxidative stress defense and DNA repair, respectively 13, 6 and 9 polymorphisms). Polymorphisms which, based on available literature, could not clearly be categorized a priori as leading to an 'increased risk' or a 'protective effect' were excluded. RESULTS The mean number of risk alleles for all investigated polymorphisms was found to be lower in the 'elderly' (17.0 ± 2.9) than the 'adolescent' (17.6 ± 3.1) subpopulation (P = 0.002). These results were not affected by gender nor smoking. The prevalence of a high (> 17 = median) number of risk alleles was less frequent in the 'elderly' (40.6%) than the 'adolescent' (51.4%) subpopulation (P = 0.002). In particular for phase II enzymes, the mean number of risk alleles was lower in the 'elderly' (4.3 ± 1.6 ) than the 'adolescent' age group (4.8 ± 1.9) P < 0.001 and the prevalence of a high (> 4 = median) number of risk alleles was less frequent in the 'elderly' (41.3%) than the adolescent subpopulation (56.3%, P < 0.001). The prevalence of a high (> 8 = median) number of risk alleles for DNA repair enzyme-coding genes was lower in the 'elderly' (37,3%) than the 'adolescent' subpopulation (45.6%, P = 0.017). CONCLUSIONS These observations are consistent with the hypothesis that, in Flanders, the prevalence of at-risk alleles in genes involved in genotoxic effects decreases with age, suggesting that persons carrying a higher number of at risk alleles (especially in phase II xenobiotic-metabolizing or DNA repair genes) are at a higher risk of morbidity and mortality from chronic diseases. Our findings also suggest that, regarding risk of disease associated with low penetrance polymorphisms, multiple polymorphisms should be taken into account, rather than single ones.
Collapse
Affiliation(s)
- Hans B Ketelslegers
- Department of Health Risk Analysis and Toxicology, Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Roger WL Godschalk
- Department of Health Risk Analysis and Toxicology, Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Ralph WH Gottschalk
- Department of Health Risk Analysis and Toxicology, Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Ad M Knaapen
- Department of Health Risk Analysis and Toxicology, Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Gudrun Koppen
- Environmental toxicology, Flemish Institute of Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium
| | - Greet Schoeters
- Environmental toxicology, Flemish Institute of Technological Research (VITO), Boeretang 200, 2400 Mol, Belgium
| | - Willy F Baeyens
- Analytical and Environmental Chemistry Department, ANCH, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Vera Nelen
- Provincial Institute of Hygiene, Kronenburgstraat 45, 2000 Antwerp, Belgium
| | - Joep PM Geraedts
- Department of Genetics & Cell Biology, Research Institute GROW, Faculty of Health, Medicine & Life Sciences, Maastricht University, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Joost HM van Delft
- Department of Health Risk Analysis and Toxicology, Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Jos CS Kleinjans
- Department of Health Risk Analysis and Toxicology, Maastricht University, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | - Nicolas A van Larebeke
- Study Centre for Carcinogenesis and Primary Prevention of Cancer, Department of Radiotherapy and Experimental Cancerology, Ghent University Hospital, De Pintelaan 185 3K3, 9000 Ghent, Belgium
| |
Collapse
|
15
|
Ari E, Kaya Y, Demir H, Cebi A, Alp HH, Bakan E, Odabasi D, Keskin S. Oxidative DNA damage correlates with carotid artery atherosclerosis in hemodialysis patients. Hemodial Int 2011; 15:453-9. [PMID: 22111813 DOI: 10.1111/j.1542-4758.2011.00568.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 05/01/2011] [Indexed: 11/28/2022]
Abstract
Oxidative stress is accepted as a nonclassical cardiovascular risk factor in chronic renal failure patients. The aim of this study was to evaluate the relation between oxidative DNA damage (8-hydroxy-2'-deoxyguanosine/deoxyguanosine [8-OHdG/dG] ratio), oxidative stress biomarkers, antioxidant enzymes, and carotid artery intima-media thickness (CIMT) in hemodialysis (HD) patients. Forty chronic HD patients without known atherosclerotic disease and 48 age- and sex-matched healthy individuals were included in the study. Plasma malondialdehyde (MDA) levels and 8-OHdG/dG ratio were determined as oxidative stress markers. Superoxide dismutase (SOD) and glutathione peroxidase (GPx) activities were measured as antioxidants. CIMT was assessed by carotid artery ultrasonography. 8-OHdG/dG ratios and MDA levels were higher; SOD and GPx activities were lower in HD patients compared to controls. HD patients had significantly higher CIMT compared to controls (0.61 ± 0.08 vs. 0.42 ± 0.05, p < 0.001). There was a significant positive correlation between CIMT and 8-OHdG/dG ratio (r = 0.57, p < 0.01) and MDA levels (r = 0.41, p < 0.01), while there was a significant negative correlation between CIMT and SOD (r = -0.47, p < 0.01) and GPx levels (r = -0.62, p < 0.01). It is firstly demonstrated that CIMT is positively correlated with oxidative DNA damage in HD patients without known atherosclerotic disease.
Collapse
Affiliation(s)
- Elif Ari
- Department of Nephrology, Van Yuksek Ihtisas Hospital, Van, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Onder G, Capoluongo E, Giovannini S, Concolino P, Russo A, Liperoti R, Bernabei R, Landi F. Interaction between GSTM1 genotype and IL-6 on mortality in older adults: results from the ilSIRENTE study. Cytokine 2010; 53:301-5. [PMID: 21169033 DOI: 10.1016/j.cyto.2010.11.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/08/2010] [Accepted: 11/23/2010] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND AIMS The inflammatory process is related to oxidative stress and inflammation was proven to be a strong determinant of the aging process and to ultimately lead to death. The aim of the present study was to assess if, in a population of older adults, the effect of antioxidant genes GSTM1 and GSTT1 genotypes on mortality may differ depending on levels of inflammation. METHODS Data are from 353 older persons aged ≥80 years enrolled in the ilSIRENTE study. Study population was divided into two groups computed based on the median value of serum IL-6 (low IL-6, n=177 and high IL-6, n=176). All participants were followed up for 48 months. RESULTS Mean age of study participants was 85.8 years (Standard Deviation 4.8), 235 (66.6%) were women. Overall 48/177 participant (27.1%) in the low IL-6 group died during the study period, compared with 97/176 (55.1%) in the high IL-6 group (p<0.001). After adjusting for potential confounders, GSTM1 wildtype had no effect on mortality in the low IL-6 group (RR=1.07; 95% CI 0.46-2.47), but it was associated with a significant lower mortality rate in the high IL-6 level (RR=0.33; 95% CI 0.15-0.69). Testing the interaction between IL-6 and GSTM1 genotype, we found a significant result (p=0.02). No significant effect of GSTT1 genotype on mortality was shown in participants with low and high IL-6 level. CONCLUSION GSTM1 wildtype is associated with reduced mortality among older adults with high levels of inflammation, but not among those with low levels of inflammation.
Collapse
Affiliation(s)
- Graziano Onder
- Centro Medicina dell'Invecchiamento, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Khelil M, Zenati A, Makrelouf M, Otmane A, Tayebi B. Polymorphisms in NAT2 gene and atherosclerosis in an Algerian population. Arch Med Res 2010; 41:215-20. [PMID: 20682180 DOI: 10.1016/j.arcmed.2010.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Accepted: 03/19/2010] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND AIMS The etiology of atherosclerosis is multifactorial. Genetic and environmental factors are involved in the development of atherosclerosis. Human arylamine N-acetyltransferase 2 (NAT2) is an important metabolizing enzyme that exhibits genetic polymorphisms and modifies individual response and/or toxicity to many xenobiotics. We undertook this study to investigate the NAT2 polymorphisms in patients with atherosclerosis. METHODS Genotyping for NAT2 alleles was performed using polymerase chain reaction-restriction fragment-length polymorphism (PCR-RFLP) in 285 Algerian patients with atherosclerosis and 286 controls. RESULTS There was no association between NAT2 polymorphisms and atherosclerosis risk. However, the haplotype NAT2(*)5F decreased susceptibility to the disease (p = 0.005, OR = 0.55, 95% CI = 0.37-0.84). The frequency of the slow acetylator phenotype was approximately 50% in both cases and controls. CONCLUSIONS These results suggest that NAT2 polymorphisms may not be involved in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Malika Khelil
- Département de Biologie Cellulaire et Moléculaire, Faculté des Sciences Biologiques, Université des Sciences et de la Technologie Houari, Boumediène, Alger, Algérie.
| | | | | | | | | |
Collapse
|
18
|
Pavanello S. Metabolic and DNA Repair Variations in Susceptibility to Genotoxins. Polycycl Aromat Compd 2010. [DOI: 10.1080/10406630308061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Sofia Pavanello
- a Section of Occupational Health, Department of Environmental Medicine and Public Health , University of Padova , Padova , Italy
| |
Collapse
|
19
|
Wang CL, Lin TH, Lin HY, Sheu SH, Yu ML, Hsiao PJ, Lin KD, Hsu C, Yang YH, Shin SJ. The 8-oxoguanine glycosylase I (hOGG1) Ser326Cys variant affects the susceptibility to multi-vessel disease in Taiwan coronary artery disease patients. Thromb Res 2010; 126:319-323. [PMID: 20667409 DOI: 10.1016/j.thromres.2010.06.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 06/20/2010] [Accepted: 06/29/2010] [Indexed: 10/19/2022]
Abstract
8-hydroxydeoxyguanosine, the key lesion of oxidative DNA damage, contributes to the development of coronary artery disease (CAD). In humans, 8-hydroxydeoxyguanosine is repaired by the enzyme 8-oxoguanine glycosylase I (hOGG1). We investigated the association between the hOGG1 Ser(326)Cys polymorphism and the presence and the severity of CAD in a Taiwan population. Genotypes of the hOGG1 Ser(326)Cys polymorphism were determined from 1397 participants enrolled in this study (378 CAD patients and 1019 controls). CAD severity was indicated both by number of vessels affected (single-vessel disease, SVD vs. multi-vessel disease, MVD), and by individual diffuse score. Real-time polymerase chain reaction was used to determine genotype, using allele-specific TaqMan probes. We found that presence of the hOGG1 Ser(326)Cys polymorphism was associated with a significantly increased risk of CAD and multi-vessel disease when assuming a dominant model of inheritance (OR: 1.52 [95%:1.082~2.133], p=0.015; OR: 2.26 [95%:1.232~4.156], p=0.007). This result was confirmed by multivariate analysis, after adjustment for age, gender, body-mass index, diabetes hypertension, hypercholesterolemia and smoking (OR: 1.78 [95%:1.127~2.806], p<0.005; OR: 2.44 [95%:1.276~4.651], p<0.001). In the present study, hOGG1 Ser(326)Cys polymorphism is a novel genetic marker to be independently associated with the development and severity of CAD in Taiwanese population.
Collapse
Affiliation(s)
- Chiao-Ling Wang
- Graduate Institute of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Taiwan, ROC
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Leopold JA, Loscalzo J. Oxidative risk for atherothrombotic cardiovascular disease. Free Radic Biol Med 2009; 47:1673-706. [PMID: 19751821 PMCID: PMC2797369 DOI: 10.1016/j.freeradbiomed.2009.09.009] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 08/31/2009] [Accepted: 09/06/2009] [Indexed: 02/07/2023]
Abstract
In the vasculature, reactive oxidant species, including reactive oxygen, nitrogen, or halogenating species, and thiyl, tyrosyl, or protein radicals may oxidatively modify lipids and proteins with deleterious consequences for vascular function. These biologically active free radical and nonradical species may be produced by increased activation of oxidant-generating sources and/or decreased cellular antioxidant capacity. Once formed, these species may engage in reactions to yield more potent oxidants that promote transition of the homeostatic vascular phenotype to a pathobiological state that is permissive for atherothrombogenesis. This dysfunctional vasculature is characterized by lipid peroxidation and aberrant lipid deposition, inflammation, immune cell activation, platelet activation, thrombus formation, and disturbed hemodynamic flow. Each of these pathobiological states is associated with an increase in the vascular burden of free radical species-derived oxidation products and, thereby, implicates increased oxidant stress in the pathogenesis of atherothrombotic vascular disease.
Collapse
Affiliation(s)
- Jane A Leopold
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
21
|
Yang Y, Parsons KK, Chi L, Malakauskas SM, Le TH. Glutathione S-transferase-micro1 regulates vascular smooth muscle cell proliferation, migration, and oxidative stress. Hypertension 2009; 54:1360-8. [PMID: 19822795 DOI: 10.1161/hypertensionaha.109.139428] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glutathione S-transferase-micro1, GSTM1, belongs to a superfamily of glutathione S-transferases that metabolizes a broad range of reactive oxygen species and xenobiotics. Across species, genetic variants that result in decreased expression of the Gstm1 gene are associated with increased susceptibility for vascular diseases, including atherosclerosis in humans. We previously identified Gstm1 as a positional candidate in our gene mapping study for susceptibility to renal vascular injury characterized by medial hypertrophy and hyperplasia of the renal vessels. To determine the role of Gstm1 in vascular smooth muscle cells (VSMCs), we isolated VSMCs from mouse aortas. We demonstrate that VSMCs from the susceptible C57BL/6 mice have reduced expression of Gstm1 mRNA and its protein product compared with that of the resistant 129 mice. After serum stimulation, C57BL/6 VSMCs proliferate and migrate at a much faster rate than 129 VSMCs. Furthermore, C57BL/6 VSMCs have higher levels of reactive oxygen species and exhibit exaggerated p38 mitogen-activated protein kinase phosphorylation after exposure to H(2)O(2). To establish causality, we show that knockdown of Gstm1 by small interfering RNA results in increased proliferation of VSMCs in a dose-dependent manner, as well as in increased reactive oxygen species levels and VSMC migration. Moreover, Gstm1 small interfering RNA causes increased p38 mitogen-activated protein kinase phosphorylation and attenuates the antiproliferative effect of Tempol. Our data suggest that Gstm1 is a novel regulator of VSMC proliferation and migration through its role in handling reactive oxygen species. Genetic variants that cause a decremental change in expression of Gstm1 may permit an environment of exaggerated oxidative stress, leading to susceptibility to vascular remodeling and atherosclerosis.
Collapse
Affiliation(s)
- Yanqiang Yang
- University of Virginia, Box 800133, Charlottesville, VA 22908, USA
| | | | | | | | | |
Collapse
|
22
|
Influence of genetic polymorphisms of glutathione S-transferase T1 (GSTT1) and M1 (GSTM1) on hematological parameters. Mol Biol Rep 2009; 37:249-53. [DOI: 10.1007/s11033-009-9662-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Accepted: 07/24/2009] [Indexed: 12/11/2022]
|
23
|
Wang LS, Tang JJ, Tang NP, Wang MW, Yan JJ, Wang QM, Yang ZJ, Wang B. Association of GSTM1 and GSTT1 gene polymorphisms with coronary artery disease in relation to tobacco smoking. Clin Chem Lab Med 2009; 46:1720-5. [PMID: 19055448 DOI: 10.1515/cclm.2008.353] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Recent studies suggest that the common variant in the glutathione S-transferase (GST) M1 (GSTM1) and T1 (GSTT1) gene is associated with the risk of smoking-related coronary artery disease (CAD). Intra-ethnic as well as inter-ethnic differences are known to impact the frequencies of GST gene polymorphisms, thus influencing its interactive effect with tobacco smoking on CAD risk. The aim of the present study was to evaluate the interaction of the genetic polymorphisms of GSTM1 and GSTT1 with cigarette smoking and the risk of CAD in a Chinese population. METHODS We conducted a study with 277 CAD patients and 277 controls matched by age and sex to examine the prevalence of GSTM1 and GSTT1 polymorphism in CAD. RESULTS We found that homozygous deletion of GSTM1 had a frequency of 32.1% among patients with CAD and 21.3% among those without CAD (p=0.004). The frequency of the GSTT1(null) genotype was 27.8% among the patients with CAD and 19.1% among CAD-free subjects (p=0.016). Patients who smoked having both the wild-type genotypes of GSTM1 and GSTT1 were protected from developing coronary heart disease (p<0.001). Moreover, smokers with combined GSTM1(null)GSTT1(null) genotypes had a significantly higher number of stenosed vessels than those with the positive genotype (p=0.02). CONCLUSIONS Our results suggest that GST polymorphisms may be a susceptibility factor to smoking-related CAD in the Chinese population.
Collapse
Affiliation(s)
- Lian-Sheng Wang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Lin YS, Hung SC, Wei YH, Tarng DC. GST M1 polymorphism associates with DNA oxidative damage and mortality among hemodialysis patients. J Am Soc Nephrol 2008; 20:405-15. [PMID: 19056870 DOI: 10.1681/asn.2008020227] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Leukocyte 8-hydroxy-2'-deoxyguanosine (8-OHdG) is a surrogate marker of oxidant-induced DNA damage in patients undergoing maintenance hemodialysis (MHD). Glutathione S-transferase M1 (GST M1) is a member of the GST family of proteins, which protect cellular DNA against oxidative damage. This study tested the association of a common GST M1 gene polymorphism [GST M1(-)], known to produce a dysfunctional enzyme, with levels of 8-OHdG in peripheral blood leukocytes and all-cause mortality among MHD patients. Among 488 MHD patients and 372 gender-matched healthy subjects, the frequency of the GST M1(-) genotype was 63.1 and 60.2%, respectively. The GST M1(-) genotype was associated with significantly higher levels of leukocyte 8-OHdG compared with the GST M1(+) genotype, even after adjustment for potential confounders (P < 0.001). Moreover, GST M1(-) patients who also had a common polymorphism in the DNA repair enzyme 8-oxoguanine DNA glycosylase 1 or who underwent dialysis with a bioincompatible cellulose membrane had the highest median levels of leukocyte 8-OHdG. Multivariate Cox regression revealed that among MHD patients, GST M1(-) genotype approximately doubled the risk for all-cause mortality (hazard ratio 2.24; 95% confidence interval 1.30 to 4.51) during the mean follow-up of 34 mo. In conclusion, patients without GST M1 activity are more vulnerable to oxidative stress and are at greater risk for death compared with those who possess GST M1 activity.
Collapse
Affiliation(s)
- Yi-Sheng Lin
- Division of Nephrology, Taipei City Hospital Zhongxiao Branch, Taipei, Taiwan
| | | | | | | |
Collapse
|
25
|
Kim SJ, Kim MG, Kim KS, Song JS, Yim SV, Chung JH. Impact of glutathione S-transferase M1 and T1 gene polymorphisms on the smoking-related coronary artery disease. J Korean Med Sci 2008; 23:365-72. [PMID: 18583868 PMCID: PMC2526535 DOI: 10.3346/jkms.2008.23.3.365] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Glutathione S-transferase (GST) plays a key role in the detoxification of xenobiotic atherogen generated by smoking. To analyze the effect of GSTM1/T1 gene polymorphisms on the development of smoking-related coronary artery disease (CAD), 775 Korean patients who underwent coronary angiography were enrolled. The subjects were classified by luminal diameter stenosis into group A (>50%), B (20-50%), or C (<20%). GSTM1 and GSTT1 gene polymorphisms were analyzed using multiplex polymerase chain reaction (PCR) for GSTM1/T1 genes and CYP1A1 gene for internal control. Of 775 subjects, 403 patients belonged to group A. They had higher risk factors for CAD than group B (N=260) and group C (N=112). The genotype frequencies of null GSTM1 and GSTT1 showed no significant differences among 3 groups. Considering the effect of GSTM1 gene polymorphisms on the smoking-related CAD, smokers with GSTM1 null genotype had more increased risk for CAD than non-smoker with GSTM1 positive genotype (odds ratios [OR], 2.07, confidence interval [CI], 1.06-4.07). Also the effect of GSTT1 gene polymorphism on smoking-related CAD showed the same tendency as GSTM1 gene (OR, 2.00, CI, 1.05-3.84). This effect of GSTM1/T1 null genotype on smoking-related CAD was augmented when both gene polymorphisms were considered simultaneously (OR, 2.76, CI, 1.17-6.52). We concluded that GSTM1/T1 null genotype contributed to the pathogenesis of smoking-related CAD to some degree.
Collapse
Affiliation(s)
- Soo-Joong Kim
- Department of Cardiology, Kyunghee University College of Medicine, Seoul, Korea.
| | | | | | | | | | | |
Collapse
|
26
|
Nair J, De Flora S, Izzotti A, Bartsch H. Lipid peroxidation-derived etheno-DNA adducts in human atherosclerotic lesions. Mutat Res 2007; 621:95-105. [PMID: 17412369 DOI: 10.1016/j.mrfmmm.2007.02.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 05/14/2023]
Abstract
Atherosclerosis and cancer are characterized by uncontrolled cell proliferation and share common risk factors, such as cigarette smoking, dietary habits and ageing. Growth of smooth muscle cells (SMCs) in atherosclerotic plaques may result from DNA damage, caused either by exogenous mutagens or by agents endogenously generated due to oxidative stress and lipid peroxidation (LPO). Hydroxy-2-nonenal (HNE), a major LPO product, binds covalently to cellular DNA to form the exocyclic etheno-DNA-base adducts, 1,N(6)-ethenodeoxyadenine (varepsilondA) and 3,N(4)-ethenodeoxycytosine (varepsilondC). By applying an ultrasensitive (32)P-postlabeling-immunoaffinity method, varepsilondA and varepsilondC were quantified in abdominal aorta SMCs from 13 atherosclerotic patients and 3 non-smoking subjects without atherosclerotic lesions. The levels of etheno-adducts ranged for varepsilondA from 2.3 to 39.6/10(8)dA and for varepsilondC from 10.7 to 157.7/10(8)dC, with a high correlation between varepsilondA and varepsilondC (r=0.84, P=0.0001). Etheno-adduct levels were higher in atherosclerotic smokers than in ex-smokers for both varepsilondA (means 15.2 versus 7.3, P=0.06) and varepsilondC (71.9 versus 51.6, not significant). varepsilondC levels were higher in either ex-smokers (P=0.03) or smokers (P=0.07) than in non-smokers. There was a poor correlation between either varepsilondA or varepsilondC and 8-hydroxy-2'-deoxyguanosine, whereas significant positive correlations were detected with the levels of several postlabeled bulky aromatic DNA adducts. In conclusion, two different types of DNA damage may be involved in atherosclerotic plaque formation and progression: (i) bulky aromatic compounds, to which aorta SMCs are chronically exposed in smokers, can either covalently bind to DNA, induce redox-cycling via quinone intermediates and/or activate local chronic inflammatory processes in the arterial wall; ii) this in turn leads to a self perpetuating generation of reactive oxygen species, LPO-products and increasing DNA-damage, as documented by the presence of high levels of miscoding etheno-DNA adducts in human aorta SMCs.
Collapse
Affiliation(s)
- Jagadeesan Nair
- Division of Toxicology and Cancer Risk Factors, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
27
|
De Flora S, Izzotti A. Mutagenesis and cardiovascular diseases Molecular mechanisms, risk factors, and protective factors. Mutat Res 2007; 621:5-17. [PMID: 17383689 DOI: 10.1016/j.mrfmmm.2006.12.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 05/14/2023]
Abstract
Although no generalization can be made, it is of interest that cancer, cardiovascular diseases, and other chronic conditions often share common risk factors and common protective factors as well as common pathogenetic determinants, such as DNA damage, oxidative stress, and chronic inflammation. Atherosclerosis is the most important cause of vascular forms representing the major cause of death in the population of many geographical areas. A great deal of studies support the "response-to-injury" theory. A variety of experimental and epidemiological findings are also in favor of the somatic mutation theory, which maintains that the earliest event in the atherogenic process is represented by mutations in arterial smooth muscle cells, akin to formation of a benign tumor. These two theories can be harmonized, also taking into account the highly diversified nature of atherosclerotic lesions. Molecular epidemiology studies performed in our laboratory and other laboratories have shown that DNA adducts are systematically present in arterial smooth muscle cells, and their levels are correlated with atherogenic risk factors known from traditional epidemiology. Oxidative DNA damage was also consistently detected in these cells. The role of glutathione S-transferase polymorphisms on the frequency of the above molecular alterations and of arterial diseases is rather controversial. Prevention of both cancer and atherosclerosis is based on avoidance of exposure to risk factors and on fortification of the host defense mechanisms by means of dietary principles and chemopreventive drugs.
Collapse
Affiliation(s)
- Silvio De Flora
- Department of Health Sciences, University of Genoa, Via A. Pastore 1, I-16132 Genoa, Italy.
| | | |
Collapse
|
28
|
|
29
|
Manfredi S, Federici C, Picano E, Botto N, Rizza A, Andreassi MG. GSTM1, GSTT1 and CYP1A1 detoxification gene polymorphisms and susceptibility to smoking-related coronary artery disease: a case-only study. Mutat Res 2007; 621:106-12. [PMID: 17408703 DOI: 10.1016/j.mrfmmm.2007.02.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 05/14/2023]
Abstract
Cigarette smoking is a powerful risk factor for coronary artery disease (CAD), leading to the formation of DNA alterations within blood vessels and heart. However, the degree of smoking-related atherosclerosis varies from individual to individual. Genetic polymorphisms of relevant xenobiotic metabolising enzymes may determine the susceptibility of an individual response to environmental toxicants. The purpose of this study was to test the hypothesis that the inheritance of polymorphic genes encoding cytochrome P450 1A1 (CYP1A1 MspI) and glutathione S-transferases (GSTM1(null) and GSTT1(null)) may be causally associated with the presence and severity of smoking-induced CAD. In a case-only design, 222 (179 male, 57.8+/-10.3 years) consecutive smoker patients who had undergone elective and diagnostic coronary angiography were recruited. We found a group (n=169) of smoker patients with significant CAD, defined as>50% reduction in diameter of at least one major vessel, and a group without obstructive CAD (n=53). No significant differences were observed in CYP1A1 genotypes frequencies between CAD and non-CAD smokers (p=0.1). Homozygous deletion of GSTM1 had a frequency of 58.6% among patients with CAD and 45.3% among those without CAD (p=0.08). The frequency of the GSTT1(null) genotype was 43.8% among the patients with CAD and 24.5% among CAD-free subjects (p=0.01). After adjustment for traditional risk factors, the presence of combined GSTM1(null)GSTT1(null) genotypes was significantly associated with an increased risk of CAD (OR=3.9; 95% CI: 1.3-11.4, p=0.01). Moreover, smokers with combined GSTM1(null)GSTT1(null) genotypes had significantly higher number of stenosed vessels than those with the positive genotype (2.3+/-0.9 versus 1.7+/-0.8, p=0.03). Our findings showed that smokers carrying GST deleted genotypes have an increased susceptibility to the smoking related coronary artery disease. Exploring gene-smoking effect provides an excellent model in order to understand gene-environment toxicants interaction and its implications to cardiovascular disease.
Collapse
Affiliation(s)
- Samantha Manfredi
- CNR Institute of Clinical Physiology, G. Pasquinucci Hospital, Massa, Italy
| | | | | | | | | | | |
Collapse
|
30
|
Izzotti A, Piana A, Minniti G, Vercelli M, Perrone L, De Flora S. Survival of atherosclerotic patients as related to oxidative stress and gene polymorphisms. Mutat Res 2007; 621:119-28. [PMID: 17383690 DOI: 10.1016/j.mrfmmm.2006.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 05/14/2023]
Abstract
A prospective molecular epidemiology study was implemented in a cohort of 98 subjects suffering from severe atherosclerotic lesions requiring removal of an abdominal aorta fragment. We previously published the results relative to detection, in the aorta medium layer, of bulky DNA adducts and fluorescent polycyclic aromatic hydrocarbon-related DNA adducts, oxidative DNA damage, and mitochondrial DNA 4977 common deletion, as well as GSTM1 and GSTT1 gene polymorphisms. We report herein new data, relative to oxidative stress biomarkers, including oxidative DNA damage in both inner and medium aorta layers, malondialdehyde in the medium layer, homocysteine and reduced glutathione in plasma, and those relative to additional gene polymorphisms, including NAT1, NAT2, OGG1, MTHFR, Leiden factor V, and prothrombin. The results of biochemical and molecular analyses were related to survival of the patients, whose average age was 70 at the start of the follow up. During the following 14 years, 71.4% of them died. The results obtained provide evidence for the crucial impact of oxidative stress and certain gene polymorphisms on clinical and biochemical patterns as well as on survival of patients. Survival was significantly affected not only by traditional risk factors for atherosclerosis but also by molecular end-points and adverse gene polymorphisms, and by their combinations.
Collapse
Affiliation(s)
- Alberto Izzotti
- Department of Health Sciences, University of Genoa, via A. Pastore 1, I-16132 Genoa, Italy.
| | | | | | | | | | | |
Collapse
|
31
|
Seedhouse C, Russell N. Advances in the understanding of susceptibility to treatment-related acute myeloid leukaemia. Br J Haematol 2007; 137:513-29. [PMID: 17539774 DOI: 10.1111/j.1365-2141.2007.06613.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Treatment-related acute myeloid leukaemia (t-AML) is a devastating complication following exposure to the cytotoxic and genotoxic agents used to treat a primary malignancy. Whilst the incidence of t-AML is rising, it still only occurs in a minority of patients who have received chemotherapy and/or radiotherapy treatment and hence it is important to identify factors that may confer susceptibility to the development of the condition. This paper reviews the literature and discusses the advances and limitations in our understanding of susceptibility factors to t-AML. In particular, it concentrates upon genetic polymorphisms in detoxification genes and in genes belonging to the major DNA repair pathways. This review also considers more novel susceptibility factors, such as those proposed to determine stem cell number. Increased understanding of t-AML susceptibility may enable steps to be taken to prevent its development and increase the effectiveness of treatment of the disease.
Collapse
Affiliation(s)
- Claire Seedhouse
- Department of Academic Haematology, University of Nottingham, Nottingham University Hospitals, Nottingham, UK.
| | | |
Collapse
|
32
|
Murgia E, Maggini V, Barale R, Rossi AM. Micronuclei, genetic polymorphisms and cardiovascular disease mortality in a nested case-control study in Italy. Mutat Res 2007; 621:113-8. [PMID: 17448506 DOI: 10.1016/j.mrfmmm.2007.02.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 11/22/2022]
Abstract
AIM To validate the predictive value of micronuclei (MN) in peripheral blood lymphocytes (PBL) and glutathione-S-transferases (GSTs) polymorphisms (GSTM1 and GSTT1) for mortality risk (MR) of cardiovascular diseases (CVD). METHODS Blood samples from 1650 healthy subjects selected from the general population were collected between June 1991 and November 1993, and slides were immediately prepared for MN assessment. The vital status, or the cause of death, was monitored for all subjects until January 2005. At the end of the follow-up, 111 deaths were recorded and 39 CVD cases were observed (age range=42-88 years). Two thousand binucleated (BN) cells/subject were scored for the MN assay and GSTs genotypes were assessed on the DNA extracted from the blood or serum samples. RESULTS A significantly higher MN frequency was recorded for the case group in comparison with the control group (n=67, Kruskall-Wallis test, p=0.006) and GSTT1 null genotype was significantly less frequent in CVD patients (chi(2)-test, p=0.036). The influence of other factors were evaluated using a unconditional logistic regression that confirmed a significant association of GSTT1 positive genotype with an increased OR for CVD (OR=6.29, 95% CI 1.32-29.95) beside a significant effect of age (OR=1.13, 95% CI 1.03-1.26 year(-1)). Finally, subjects with an higher MN frequency showed a higher MR for CVD (Log-rank test, p=0.001). CONCLUSIONS MN confirmed to be a suitable cytogenetic biomarker for early prediction of CVD death. The GSTT1 positive genotype is associated with an increased MR for CVD.
Collapse
Affiliation(s)
- Elena Murgia
- Department of Biology, Pisa University, Pisa, Italy
| | | | | | | |
Collapse
|
33
|
Knaapen AM, Curfs DM, Pachen DM, Gottschalk RW, de Winther MPJ, Daemen MJ, Van Schooten FJ. The environmental carcinogen benzo[a]pyrene induces expression of monocyte-chemoattractant protein-1 in vascular tissue: a possible role in atherogenesis. Mutat Res 2007; 621:31-41. [PMID: 17376491 DOI: 10.1016/j.mrfmmm.2006.12.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2006] [Revised: 12/12/2006] [Accepted: 12/13/2006] [Indexed: 01/07/2023]
Abstract
Exposure to carcinogenic polycyclic aromatic hydrocarbons (PAHs) has been implicated in the aetiology of atherosclerosis. Previously we showed that chronic exposure of ApoE-/- mice to the prototype PAH benzo[a]pyrene (B[a]P) causes enhanced progression of atherosclerosis, which was characterised by an increased inflammatory cell content in the atherosclerotic plaques. The aim of the present study was to evaluate the effect of B[a]P on vascular expression of monocyte-chemoattractant protein 1 (MCP-1), which is a crucial molecule promoting the recruitment of monocytes into atherosclerotic lesions. We hypothesised that B[a]P-induced expression of MCP-1 is mediated through aryl hydrocarbon receptor (AhR) activation. Initially we performed in vivo studies showing that acute treatment with B[a]P induces MCP-1 gene expression in aortic tissue of ApoE-/- mice. These observations could be confirmed by in vitro studies with human endothelial cells (RF24 cell line and primary HUVEC), showing a dose- and time-dependent increase in MCP-1 expression upon exposure to B[a]P. This was paralleled by an induction of cytochrome P450 1A1 and 1B1, indicating Ah receptor activation. No increased gene expression (MCP-1, CYP1A1 and 1B1) was found upon incubation with the structural isomer benzo[e]pyrene, which is a weak AhR agonist. Moreover, B[a]P-induced MCP-1 gene and protein expression was inhibited by co-treatment with the AhR antagonist alpha-naphthoflavone. In addition to its effect on basal gene expression, we showed that B[a]P significantly enhanced TNFalpha-induced expression of MCP-1. We were unable to block B[a]P-induced MCP-1 expression by antioxidant treatment. In contrast, we found that addition of N-acetylcysteine or vitamin C enhanced transcription of MCP-1 by B[a]P. In conclusion, our studies revealed potent vascular pro-inflammatory effects of B[a]P, as evidenced by AhR-mediated induction of MCP-1. These observations further contribute to the concept that induction of inflammation is a crucial process in PAH-enhanced atherogenesis.
Collapse
Affiliation(s)
- Ad M Knaapen
- Department of Health Risk Analysis and Toxicology, Nutrition and Toxicology Research Institute Maastricht (NUTRIM), The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Mitochondria have long been known to play a critical role in maintaining the bioenergetic status of cells under physiological conditions. It was also recognized early in mitochondrial research that the reduction of oxygen to generate the free radical superoxide occurs at various sites in the respiratory chain and was postulated that this could lead to mitochondrial dysfunction in a variety of disease states. Over recent years, this view has broadened substantially with the discovery that reactive oxygen, nitrogen, and lipid species can also modulate physiological cell function through a process known as redox cell signaling. These redox active second messengers are formed through regulated enzymatic pathways, including those in the mitochondrion, and result in the posttranslational modification of mitochondrial proteins and DNA. In some cases, the signaling pathways lead to cytotoxicity. Under physiological conditions, the same mediators at low concentrations activate the cytoprotective signaling pathways that increase cellular antioxidants. Thus, it is critical to understand the mechanisms by which these pathways are distinguished to develop strategies that will lead to the prevention of cardiovascular disease. In this review, we describe recent evidence that supports the hypothesis that mitochondria have an important role in cell signaling, and so contribute to both the adaptation to oxidative stress and the development of vascular diseases.
Collapse
Affiliation(s)
- Jessica Gutierrez
- Department of Physiology and Biophysics, Center for Free Radical Biology, University of Alabama at Birmingham, USA
| | | | | | | |
Collapse
|
35
|
Van Vickle-Chavez SJ, Tung WS, Absi TS, Ennis TL, Mao D, Cobb JP, Thompson RW. Temporal changes in mouse aortic wall gene expression during the development of elastase-induced abdominal aortic aneurysms. J Vasc Surg 2006; 43:1010-20. [PMID: 16678698 DOI: 10.1016/j.jvs.2006.01.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 01/06/2006] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To characterize temporal changes in mouse aortic wall gene expression associated with the development of experimental abdominal aortic aneurysms. METHODS C57BL/6 mice underwent transient perfusion of the abdominal aorta with either elastase (n = 61) or heat-inactivated elastase as a control (n = 68). Triplicate samples of radiolabeled aortic wall complementary DNA were prepared at intervals of 0, 3, 7, 10, and 14 days, followed by hybridization to nylon microarrays (1181 genes). Autoradiographic intensity data were normalized by conversion to z scores, and differences in gene expression were defined by two-tailed z tests at a significance threshold of P < .01. RESULTS Elastase perfusion caused a progressive increase in aortic diameter up to 14 days accompanied by transmural inflammation and destructive remodeling of the elastic media. No aneurysms occurred in the control group. Compared with healthy aorta, 336 genes exhibited significant alterations during at least 1 interval after elastase perfusion (135 at more than 1 interval and 14 at all intervals), with pronounced increases for interleukin 6, cyclin E2, interleukin 1beta, osteopontin, CD14/lipopolysaccharide receptor, P-selectin glycoprotein ligand 1, and gelatinase B/matrix metalloproteinase 9 (all >20-fold on day 3). Sixty-two genes exhibited synchronous alterations in the elastase and control groups, thus suggesting a nonspecific response. By direct comparisons between the elastase and control groups, there were 384 genes with significant differences in expression for at least 1 interval after aortic perfusion, including 234 with differential upregulation (eg, p44MAPK/ERK1, osteopontin, heat shock protein 84, hypoxia-inducible factor 1alpha, apolipoprotein E, monocyte chemotactic protein 3, MIG (monokine induced by gamma interferon), and interleukin 2 receptor gamma) and 163 with differential downregulation (eg, prothrombin, granzyme B, ataxia telangiectasia mutated, and interleukin-converting enzyme). CONCLUSIONS Development of elastase-induced abdominal aortic aneurysms in mice is accompanied by altered aortic wall expression of genes associated with acute and chronic inflammation, matrix degradation, and vascular tissue remodeling. Knowledge of these alterations will facilitate further studies on the functional molecular mechanisms that underlie aneurysmal degeneration.
Collapse
|
36
|
Pernice F, Floccari F, Caccamo C, Belghity N, Mantuano S, Pacilè ME, Romeo A, Nostro L, Barillà A, Crascì E, Frisina N, Buemi M. Chromosomal damage and atherosclerosis. A protective effect from simvastatin. Eur J Pharmacol 2006; 532:223-9. [PMID: 16483569 DOI: 10.1016/j.ejphar.2006.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2005] [Revised: 11/30/2005] [Accepted: 01/10/2006] [Indexed: 12/01/2022]
Abstract
In uremic patients, the frequency of sister chromatid exchanges appears markedly higher than in the general population. Statins are well known for their pleiotropic effects, which are independent of any reduction in cholesterol circulating levels. The aim of the present study was to determine the effects of exposure to escalating doses of simvastatin on the sister chromatid exchange rate in cultured lymphocytes in order to identify the influence of statin on genomic damage. Peripheral lymphocytic samples for culture were obtained from 25 healthy volunteers, 20 patients with documented carotid atherosclerosis and 30 atherosclerotic patients on maintenance regular acetate-free biofiltration. Hemodialyzed patients had a greater percentage of high frequency cells (50%) than healthy controls (3%) and a significantly higher average number of sister chromatid (9.82+/-2.1 vs. 4.65+/-2.18). The subgroup of hemodialyzed patients with high plaque score values was characterized by significantly greater values for both sister chromatid exchanges rate and high frequency cells percentage. Our findings demonstrate that there is an association between sister chromatid exchanges and high frequency cells rate and atherosclerosis in acetate-free biofiltration patients. In cultures with added simvastatin, high frequency cells percentages and mean sister chromatid exchanges levels were significantly lower than in cultures with an added vehicle alone, the reduction occurring in a dose-dependent fashion, above all in cultures from end stage renal disease patients. The findings, moreover, demonstrate new effects of simvastatin, which appeared to mitigate the expression of genomic damage in our model. However, it is not yet clear whether this effect is due to the prevention of genomic damage or to the potentiation of the DNA repair capacity. Statins may therefore have an anti-atherogenic action partly ascribable to their ability to provide protection against the development of atherosclerotic plaque.
Collapse
Affiliation(s)
- Franco Pernice
- Department of Internal Medicine, University of Messina, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Voho A, Impivaara O, Järvisalo J, Metsola K, Vainio H, Hirvonen A. Distribution of glutathione S-transferase M1, P1 and T1 genotypes in different age-groups of Finns without diagnosed cancer. ACTA ACUST UNITED AC 2006; 30:144-51. [PMID: 16638627 DOI: 10.1016/j.cdp.2005.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2005] [Indexed: 11/20/2022]
Abstract
BACKGROUND Xenobiotic metabolizing enzymes (XMEs) are important detoxifiers of hazardous environmental agents, and their polymorphisms may therefore modify the risk of environmentally induced cancers. Consequently, the XME polymorphisms have been extensively studied in this context during recent years. Particular attention has been given to the polymorphisms of glutathione S-transferase (GST) M1, P1 and T1 genes. Previous studies have provided abundant data indicating these polymorphisms as important modifiers of individual susceptibility to cancers of environmental origin. It can be postulated that if the at-risk genotypes of these genes were real risk factors for the environmental cancers, their prevalence would presumably decrease with age in cancer-free part of the population. METHODS We tested the hypothesis in a population based group of 2105 Finns (1,051 men, 1,054 women) in five age strata (27, 37, 47, 57 and 67 years of age), all without clinically diagnosed cancer. RESULTS For GSTM1 genotype, a significant interaction was seen between gender and age among never smokers (p=0.003). Currently smoking men tended to be less likely (OR 0.57, 95% CI 0.31-1.03), and currently smoking women more likely (OR 1.70, 95% CI 0.97-2.97) homozygotes for the GSTP1*B allele compared with never smokers. Moreover, the likelihood of being a concurrent carrier of the putatively protective genotypes of all of the three studied GSTs was almost three-fold (OR 2.80, 95% CI 1.10-7.12) in heavy smokers in the two oldest age-groups compared with the other genotypes. CONCLUSIONS Our findings based on a novel study design provide support to the previous case-control studies suggesting that GST genotypes modify individual risk of environmentally-induced cancers.
Collapse
Affiliation(s)
- Anu Voho
- Finnish Institute of Occupational Health, Topeliuksenkatu 41 a A, 41 a A, FI-00250, Helsinki, Finland
| | | | | | | | | | | |
Collapse
|
38
|
Cooke MS, Olinski R, Evans MD. Does measurement of oxidative damage to DNA have clinical significance? Clin Chim Acta 2005; 365:30-49. [PMID: 16214123 DOI: 10.1016/j.cca.2005.09.009] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Revised: 09/11/2005] [Accepted: 09/11/2005] [Indexed: 12/31/2022]
Abstract
Oxidative damage to DNA is the seemingly inevitable consequence of cellular metabolism. Furthermore, despite protective mechanisms, cellular levels of damage may increase under conditions of oxidative stress, arising from exposure to a variety of physical or chemical insults. Elevated levels of oxidatively damaged DNA have been measured in numerous diseases, and as a result, it has been hypothesised that such damage plays an integral role in the aetiology of that disease. This review examines the validity of this hypothesis, exploring the mechanisms by which oxidative DNA damage may lead to disease. We conclude that further validation of biomarkers of oxidative DNA damage, along with further elucidation of the role of damage in disease, may allow these biomarkers to become potentially useful clinical tools.
Collapse
Affiliation(s)
- Marcus S Cooke
- Radiation and Oxidative Stress Group, Department of Cancer Studies and Molecular Medicine, University of Leicester, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, LE2 7LX, UK.
| | | | | |
Collapse
|
39
|
Abstract
In the vasculature, reactive oxygen species (ROS) generated by both mitochondrial respiration and enzymatic sources serve as integral components of cellular signaling and homeostatic mechanisms. Because ROS are highly reactive biomolecules, the cellular redox milieu is carefully maintained by small-molecule antioxidants and antioxidant enzymes to prevent the deleterious consequences of ROS excess. When this redox balance is perturbed, because of either increased ROS production or decreased antioxidant capacity, oxidant stress is increased in the vessel wall and, if not offset, vascular dysfunction ensues. A number of heritable polymorphisms of pro-oxidant enzymes, including 5-lipoxygenase, cyclooxygenase-2, nitric oxide synthase-3, and NAD(P)H oxidase, have been identified and found to modulate ROS production and, thereby, the risk of atherothrombotic cardiovascular disease in individuals with these genetic polymorphisms. Similarly, heritable deficiency of the antioxidant enzymes catalase, glutathione peroxidases, glutathione-
S
-transferases, heme oxygenase, and glucose-6-phosphate dehydrogenase favors ROS accumulation, and has been associated with an increased risk of vascular disease. Individually, each of these polymorphisms imposes a state of uncompensated oxidant stress on the vasculature and collectively comprise the oxidative enzymopathies.
Collapse
Affiliation(s)
- Jane A Leopold
- Whitaker Cardiovascular Institute and Evans Department of Medicine, Boston University School of Medicine, Boston, Mass 02118, USA
| | | |
Collapse
|
40
|
Curfs DMJ, Knaapen AM, Pachen DMFA, Gijbels MJJ, Lutgens E, Smook MLF, Kockx MM, Daemen MJAP, van Schooten FJ. Polycyclic aromatic hydrocarbons induce an inflammatory atherosclerotic plaque phenotype irrespective of their DNA binding properties. FASEB J 2005; 19:1290-2. [PMID: 15939734 DOI: 10.1096/fj.04-2269fje] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Although it has been demonstrated that carcinogenic environmental polycyclic aromatic hydrocarbons (PAHs) cause progression of atherosclerosis, the underlying mechanism remains unclear. In the present study, we aimed to investigate whether DNA binding events are critically involved in the progression of PAH-mediated atherogenesis. Apolipoprotein E knockout mice were orally (24 wk, once/wk) exposed to 5 mg/kg benzo[a]pyrene (B[a]P), or its nonmutagenic, noncarcinogenic structural isoform benzo[e]pyrene (B[e]P). 32P-postlabeling of lung tissue confirmed the presence of promutagenic PAH-DNA adducts in B[a]P-exposed animals, whereas in B[e]P-exposed and vehicle control animals, these adducts were undetectable. Morphometrical analysis showed that both B[a]P and B[e]P caused an increase in plaque size, whereas location or number of plaques was unaffected. Immunohistochemistry revealed no differences in oxidative DNA damage (8-OHdG) or apoptosis in the plaques. Also plasma lipoprotein levels remained unchanged after PAH-exposure. However, T lymphocytes were increased > or =2-fold in the plaques of B[a]P- and B[e]P-exposed animals. Additionally, B[a]P and to a lesser extent B[e]P exposure resulted in increased TGFbeta protein levels in the plaques, that was mainly localized in the plaque macrophages. In vitro studies using the murine macrophage like RAW264.7 cells showed that inhibition of TGFbeta resulted in decreased tumor necrosis factor (TNF) alpha release, suggesting that enhanced TGFbeta expression in the plaque macrophages contributes to the proinflammatory effects in the vessel wall. In general, this inflammatory reaction in the plaques appeared to be a local response since peripheral blood cell composition (T cells, B cells, granulocytes, and macrophages) was not changed upon PAH exposure. In conclusion, we showed that both B[a]P and B[e]P cause progression of atherosclerosis, irrespective of their DNA binding properties. Moreover, our data revealed a possible novel mechanism of PAH-mediated atherogenesis, which likely involves a TGFbeta-mediated local inflammatory reaction in the vessel wall.
Collapse
Affiliation(s)
- Daniëlle M J Curfs
- Department of Health Risk Analysis and Toxicology, University of Maastricht, Maastricht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Arvanitis DA, Flouris GA, Spandidos DA. Genomic rearrangements on VCAM1, SELE, APEG1and AIF1 loci in atherosclerosis. J Cell Mol Med 2005; 9:153-9. [PMID: 15784173 PMCID: PMC6741330 DOI: 10.1111/j.1582-4934.2005.tb00345.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The inflammatory nature of atherosclerosis has been well established. However, the initial steps that trigger this response in the arterial intima remain obscure. Previous studies reported a significant rate of genomic alterations in human atheromas. The accumulation of genomic rearrangements in vascular endothelium and smooth muscle cells may be important for disease development. To address this issue, 78 post-mortem obtained aortic atheromas were screened for microsatellite DNA alterations versus correspondent venous blood. To evaluate the significance of these observations, 33 additional histologically normal aortic specimens from age and sex-matched cases were examined. Loss of heterozygosity (LOH) was found in 47,4% of the cases and in 18,2% of controls in at least one locus. The LOH occurrence in aortic tissue is associated to atherosclerosis risk (OR 4,06, 95% CI 1,50 to 10,93). Significant genomic alterations were found on 1p32-p31, 1q22-q25, 2q35 and 6p21.3 where VCAM1, SELE, APEG1 and AIF1 genes have been mapped respectively. Our data implicate somatic DNA rearrangements, on loci associated to leukocyte adhesion, vascular smooth muscle cells growth, differentiation and migration, to atherosclerosis development as an inflammatory condition.
Collapse
Affiliation(s)
- D A Arvanitis
- Department of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | | | | |
Collapse
|
42
|
Affiliation(s)
- Friedrich C Luft
- Franz Volhard Clinic, HELIOS Kliniken Berlin, Medical Faculty of the Charité, Humboldt University, Wiltbergstrasse 50, 13125 Berlin-Buch, Germany.
| |
Collapse
|
43
|
Influence of polymorphism of glutathione S-transferase M1 on systolic blood pressure of normotensive individuals. Biochem Biophys Res Commun 2005; 326:449-54. [PMID: 15582598 DOI: 10.1016/j.bbrc.2004.11.050] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2004] [Indexed: 10/26/2022]
Abstract
Studies have indicated that systolic (SBP) and diastolic (DBP) blood pressure are multi-factorial traits and significantly heritable. The aims of the present study are to assess whether the glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) genotypes are associated with SBP and DBP of normotensive subjects and to ascertain whether the level of SBP and DBP given exposure to cigarette smoking is modified by the specific genetic polymorphisms of GSTM1 and GSTT1. This cross-sectional study was conducted on 140 subjects (49 females and 91 males) (mean age+/-SD: 38.7+/-14.7). The genotypes were determined using a polymerase chain reaction based method. Individuals were stratified according to the mean values of DBP and SBP, lower than or maximally same as the mean value defines as group I and higher than the mean value defines as group II. The logistic regression analyses were used. The best models fitted by logistic regression analysis for variables were associated with SBP and DBP. For analysis the combination of genotypes, sex, and smoking behavior was used as qualitative variables, and age, body mass index (BMI), and heart rate were used as covariates. Combination of "present-GSTT1, null-GSTM1" genotype (OR=0.001, 95% CI=0.00-0.439, P=0.025), heart rate (OR=1.065, 95% CI=1.018-1.114, P=0.006), and interaction between BMI and combination of "present-GSTT1, null-GSTM1" (OR=1.319, 95% CI=1.058-1.644, P=0.014) was associated with SBP. There was no association between either combination genotypes of GSTs or interaction of genotypes and smoking behavior on DBP. The present results suggest that the GSTM1 gene is one of the candidate genes that alter the baseline of SBP in normotensive individuals.
Collapse
|
44
|
Evans MD, Dizdaroglu M, Cooke MS. Oxidative DNA damage and disease: induction, repair and significance. MUTATION RESEARCH/REVIEWS IN MUTATION RESEARCH 2004; 567:1-61. [PMID: 15341901 DOI: 10.1016/j.mrrev.2003.11.001] [Citation(s) in RCA: 878] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2003] [Revised: 11/12/2003] [Accepted: 11/12/2003] [Indexed: 04/08/2023]
Abstract
The generation of reactive oxygen species may be both beneficial to cells, performing a function in inter- and intracellular signalling, and detrimental, modifying cellular biomolecules, accumulation of which has been associated with numerous diseases. Of the molecules subject to oxidative modification, DNA has received the greatest attention, with biomarkers of exposure and effect closest to validation. Despite nearly a quarter of a century of study, and a large number of base- and sugar-derived DNA lesions having been identified, the majority of studies have focussed upon the guanine modification, 7,8-dihydro-8-oxo-2'-deoxyguanosine (8-OH-dG). For the most part, the biological significance of other lesions has not, as yet, been investigated. In contrast, the description and characterisation of enzyme systems responsible for repairing oxidative DNA base damage is growing rapidly, being the subject of intense study. However, there remain notable gaps in our knowledge of which repair proteins remove which lesions, plus, as more lesions identified, new processes/substrates need to be determined. There are many reports describing elevated levels of oxidatively modified DNA lesions, in various biological matrices, in a plethora of diseases; however, for the majority of these the association could merely be coincidental, and more detailed studies are required. Nevertheless, even based simply upon reports of studies investigating the potential role of 8-OH-dG in disease, the weight of evidence strongly suggests a link between such damage and the pathogenesis of disease. However, exact roles remain to be elucidated.
Collapse
Affiliation(s)
- Mark D Evans
- Oxidative Stress Group, Department of Clinical Biochemistry, University of Leicester, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, LE2 7LX, UK
| | | | | |
Collapse
|
45
|
Mulvihill ER, Jaeger J, Sengupta R, Ruzzo WL, Reimer C, Lukito S, Schwartz SM. Atherosclerotic Plaque Smooth Muscle Cells Have a Distinct Phenotype. Arterioscler Thromb Vasc Biol 2004; 24:1283-9. [PMID: 15142862 DOI: 10.1161/01.atv.0000132401.12275.0c] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Objective—
The present study addresses the question, “Are plaque smooth muscles cells (SMCs) genetically distinct from medial SMCs as reflected by the ability to maintain a distinctive expression phenotype in vitro?”
Methods and Results—
Multiple cell strains were developed from carotid endarcterectomy specimens, and quadruplicate array hybridizations were completed for each sample. A new normalization protocol was developed and used to analyze the data. Permutation analysis suggests that most of the significant differences in expression could not have occurred by chance. A broad pattern of significant expression differences, consisting of almost 5% of the genes probed, was detected. Quantitative polymerase chain reaction (QPCR) confirmation was found in 70% of a subset of genes selected for validation.
Conclusions—
The SMC cultures were nearly indistinguishable by morphological features, population doubling time, and sensitivity to cell death induced by Fas cross-linking. Surprisingly, array expression analysis identified differences so extensive that we conclude that plaque and medial SMCs are distinctly different SMC cell types.
Collapse
Affiliation(s)
- Eileen R Mulvihill
- Department of Pathology, University of Washington, Box 357335, Seattle, Wash 98195-7335, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Izzotti A, Saccà SC. Glutathione S-transferase M1 and its implications in glaucoma pathogenesis: a controversial matterDOI of the original article: 10.1016/S0014-4835(03)00109-X. Exp Eye Res 2004; 79:141-2; author reply 143. [PMID: 15183109 DOI: 10.1016/j.exer.2004.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2003] [Accepted: 03/16/2004] [Indexed: 11/20/2022]
|
47
|
Masetti S, Botto N, Manfredi S, Colombo MG, Rizza A, Vassalle C, Clerico A, Biagini A, Andreassi MG. Interactive effect of the glutathione S-transferase genes and cigarette smoking on occurrence and severity of coronary artery risk. J Mol Med (Berl) 2003; 81:488-94. [PMID: 12811412 DOI: 10.1007/s00109-003-0448-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2003] [Accepted: 05/05/2003] [Indexed: 11/27/2022]
Abstract
Cardiovascular diseases and cancer are the main causes of death in developed countries. Mortality trends for these diseases suggest that they share common pathogenetic mechanisms. Glutathione S-transferase (GST) is a family of enzymes that detoxify reactive electrophiles, particularly present in tobacco smoke. Glutathione S-transferase null M1 and T1 (GSTM1 and GSTT1) genotypes have often been associated with increased risk of developing cancer. Our hypothesis was that the polymorphic GSTM1 and GSTT1 genes modulate the risk of smoking-coronary artery disease (CAD). We evaluated the distribution of GST genotypes in 430 angiographically defined patients (308 CAD and 122 non-CAD). The frequencies of GST null genotypes did not differ significantly between patients with CAD and without CAD. However, smokers with GSTM1 and GSTT1 null genotypes had a significantly higher risk of CAD than never-smokers with these genotypes present (OR 2.2 and 3.4 for smokers with null GSTM1 and GSTT1 genes, respectively). There was also evidence of multiple interaction between GSTM1 and GSTT1 deleted genotypes and smoking. In nonsmokers carrying both null genotypes the risk of CAD was 0.66. In smokers with both present genotypes the OR was 1.5 and was significantly increased in smokers with concurrent lack for GSTM1 and GSTT1 genes (OR=4.0). Moreover, smokers lacking GST genes had both more stenosed vessels and a higher Duke score than smokers expressing the genes. We also examined the levels of DNA damage in 66 men patients using the micronucleus test, a sensitive assay for evaluating chromosome damage. Micronucleus levels were higher in smokers with null genes than in smokers with present genes. These observations suggest that GST-null genotypes strengthen the effect of smoking on CAD risk by modulating the detoxification of genotoxic atherogens.
Collapse
|
48
|
Abstract
Oxidative DNA damage is an inevitable consequence of cellular metabolism, with a propensity for increased levels following toxic insult. Although more than 20 base lesions have been identified, only a fraction of these have received appreciable study, most notably 8-oxo-2'deoxyguanosine. This lesion has been the focus of intense research interest and been ascribed much importance, largely to the detriment of other lesions. The present work reviews the basis for the biological significance of oxidative DNA damage, drawing attention to the multiplicity of proteins with repair activities along with a number of poorly considered effects of damage. Given the plethora of (often contradictory) reports describing pathological conditions in which levels of oxidative DNA damage have been measured, this review critically addresses the extent to which the in vitro significance of such damage has relevance for the pathogenesis of disease. It is suggested that some shortcomings associated with biomarkers, along with gaps in our knowledge, may be responsible for the failure to produce consistent and definitive results when applied to understanding the role of DNA damage in disease, highlighting the need for further studies.
Collapse
Affiliation(s)
- Marcus S Cooke
- Oxidative Stress Group, Department of Clinical Biochemistry, University of Leicester, Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, LE2 7LX, UK.
| | | | | | | |
Collapse
|
49
|
Abstract
BACKGROUND Little is known about the molecular mechanisms responsible for the development of glaucoma, the leading cause of irreversible blindness worldwide. Some investigators have hypothesized that oxidative damage may be involved. We evaluated oxidative deoxyribonucleic acid (DNA) damage, in terms of 8-hydroxy-2'-deoxyguanosine (8-OH-dG), in the eyes of glaucoma patients. METHODS Levels of 8-OH-dG were measured in the trabecular meshwork region from 42 patients with glaucoma and 45 controls of similar age and sex. Genotypes of glutathione S-transferase isoenzymes (GSTM1 and GSTT1) were assessed by polymerase chain reaction in the same DNA samples. RESULTS Levels of 8-OH-dG were significantly higher in glaucoma patients than in controls. Oxidative DNA damage in patients with glaucoma correlated significantly with intraocular pressure; in patients with primary open-angle glaucoma, it also correlated with visual field defects. GSTT1 was similar in the two groups, and had no effect on 8-OH-dG levels. Conversely, 8-OH-dG levels were significantly higher in GSTM1-null than in GSTM1-positive subjects. The GSTM1-null genotype was significantly more common in patients with primary open-angle glaucoma than in controls. CONCLUSION Oxidative DNA damage is significantly increased in the trabecular meshwork of glaucoma patients. GSTM1 gene deletion, which has been associated with an increased risk of cancer at various sites and molecular lesions in atherosclerosis, predisposes to more severe oxidative DNA damage in glaucoma patients. These findings may contribute to understanding the pathogenesis of glaucoma and may be useful in the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Alberto Izzotti
- Department of Health Sciences, University of Genoa, Via A. Pastore 1, I-16132 Genoa, Italy
| | | | | | | |
Collapse
|
50
|
Wilson MH, Grant PJ, Kain K, Warner DP, Wild CP. Association between the risk of coronary artery disease in South Asians and a deletion polymorphism in glutathione S-transferase M1. Biomarkers 2003; 8:43-50. [PMID: 12519635 DOI: 10.1080/1354750021000042439] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
South Asians living in Western societies show a greater risk of coronary artery disease (CAD) than the indigenous Caucasian population, probably related to the change to a Westernised lifestyle and an associated genetic susceptibility. Modulation of DNA damage and mutation caused by polymorphisms in detoxification enzymes, including the glutathione S-transferases (GSTs), is a well-established risk factor for tobacco-related carcinogenesis, and a similar change in cellular damage may be involved in the risk of vascular disease associated with tobacco smoking. In this study we examined whether polymorphisms in GST genes influence the risk of CAD in a case-control group of South Asians, following our recent observation of such an association in Caucasians from the same region of the UK. Blood was obtained from 170 patients of South Asian origin admitted for angiographic investigation of chest pain and from 203 controls. Patients were subdivided into those with and without previous acute myocardial infarction (AMI), and DNA was analysed for deletions in the GSTM1 and GSTT1 genes. An association was found between the prevalence of the GSTM1 null genotype and the risk of developing CAD in this study population. The frequency of the null genotype was 52.7% in healthy controls and 41.2% in patients (odds ratio [OR] 0.63, 95% confidence interval [95% CI] 0.42-0.95, p = 0.029). The effect was similar in subjects with or without a prior history of AMI. The association was also independent of smoking history, with both non-smokers and smokers showing a similar pattern of genotype distribution, the frequency of the null genotype being 51.2% in controls versus 37.0% in patients in 'never' smokers (OR 0.56, 95% CI 0.33-0.94, p = 0.037) and 60.0% in controls versus 46.2% in patients in 'ever' smokers (OR 0.57, 95% CI 0.25-1.28, p = 0.223). The association remained after adjusting for age, sex, body mass index and the presence or absence of stenosis. No significant associations were observed between the GSTT1 genotype and cardiovascular disease (chi(2) test, p > 0.1). The results of this study indicate that the GSTM1 null genotype is protective against both CAD and AMI. However, further study is required in order to elucidate the, as yet unexplained, mechanisms underlying this association.
Collapse
Affiliation(s)
- Michael H Wilson
- Molecular Epidemiology Unit, Epidemiology and Health Services Research, University of Leeds, Leeds, UK
| | | | | | | | | |
Collapse
|